32 results on '"Eugene Lifshits"'
Search Results
2. Supplementary Figures 1-10, Tables 1-2 from BIM Expression in Treatment-Naïve Cancers Predicts Responsiveness to Kinase Inhibitors
- Author
-
Jeffrey A. Engelman, Rakesh K. Jain, Dora Dias-Santagata, Miguel N. Rivera, Carlos L. Arteaga, José Baselga, Henry L. Gómez, Cyril H. Benes, Erik J. Coffman, Sylvie Roberge, Eugene Lifshits, Alona Muzikansky, Youngchul Song, Sarah F. Pollack, Subba R. Digumarthy, Joao Incio, Euiheon Chung, Belinda A. Waltman, Lecia V. Sequist, Hiromichi Ebi, Ryan B. Corcoran, and Anthony C. Faber
- Abstract
PDF file - 21944K
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figures 1 through 3 from Spontaneous Reversion of the Angiogenic Phenotype to a Nonangiogenic and Dormant State in Human Tumors
- Author
-
George N. Naumov, Judah Folkman, Randolph S. Watnick, Lars A. Akslen, Oddbjorn Straume, Karl-Henning Kalland, Kamila Naxerova, Soo-Young Kang, Michael Lampa, Elise R. Bender, Anne M. Oyan, Trond H. Bø, Eugene Lifshits, Anna Blois, Lorna M. Cryan, David Zurakowski, Katherine Novak, and Michael S. Rogers
- Abstract
PDF - 254K, Supplemental Figure 1: In vivo proliferation of angiogenic and non-angiogenic cell clones. Supplemental Figure 2: Chromosomal locations of genes with relative overexpression in either angiogenic or revertant clones. Supplemental Figure 3: Canonical pathways overrepresented in angiogenic and revertant clones.
- Published
- 2023
- Full Text
- View/download PDF
4. Interview with Dr. Engelman from BIM Expression in Treatment-Naïve Cancers Predicts Responsiveness to Kinase Inhibitors
- Author
-
Jeffrey A. Engelman, Rakesh K. Jain, Dora Dias-Santagata, Miguel N. Rivera, Carlos L. Arteaga, José Baselga, Henry L. Gómez, Cyril H. Benes, Erik J. Coffman, Sylvie Roberge, Eugene Lifshits, Alona Muzikansky, Youngchul Song, Sarah F. Pollack, Subba R. Digumarthy, Joao Incio, Euiheon Chung, Belinda A. Waltman, Lecia V. Sequist, Hiromichi Ebi, Ryan B. Corcoran, and Anthony C. Faber
- Abstract
mp3 file (8.8 MB). In the September edition of the Cancer Discovery podcast, Executive Editor Mark Landis talks with Jeffrey A. Engelman about his paper, which suggests that quantitation of pretreatment RNA levels of the pro-apoptotic factor BIM can predict the efficacy of tyrosine kinase inhibitor therapy in oncogene-addicted cancers.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Data from EML4-ALK Fusion Gene and Efficacy of an ALK Kinase Inhibitor in Lung Cancer
- Author
-
Pasi A. Jänne, Matthew Meyerson, Charles Lee, Nathanael S. Gray, Jeffrey A. Engelman, J. Paul Marcoux, Neal Lindeman, Christopher Ducko, David J. Sugarbaker, William G. Richards, Jinseon Lee, Roman Thomas, Derek Chiang, Jhingook Kim, Hwan Geun Choi, Alison J. Holmes, Eugene Lifshits, Carly Murphy, Kreshnik Zejnullahu, Craig Mermel, and Jussi P. Koivunen
- Abstract
Supplementary Data from EML4-ALK Fusion Gene and Efficacy of an ALK Kinase Inhibitor in Lung Cancer
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Materials and Methods from Failure to Induce Apoptosis via BCL-2 Family Proteins Underlies Lack of Efficacy of Combined MEK and PI3K Inhibitors for KRAS-Mutant Lung Cancers
- Author
-
Jeffrey A. Engelman, Kwok-Kin Wong, Mari Mino-Kenudson, Rebecca S. Heist, Anthony Letai, Kristopher A. Sarosiek, Zandra Walton, Katherine A. Cheng, Zhao Chen, Eugene Lifshits, Anthony C. Faber, Alan Yeo, and Aaron N. Hata
- Abstract
PDF file - 94KB
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figures 1 - 19 from Failure to Induce Apoptosis via BCL-2 Family Proteins Underlies Lack of Efficacy of Combined MEK and PI3K Inhibitors for KRAS-Mutant Lung Cancers
- Author
-
Jeffrey A. Engelman, Kwok-Kin Wong, Mari Mino-Kenudson, Rebecca S. Heist, Anthony Letai, Kristopher A. Sarosiek, Zandra Walton, Katherine A. Cheng, Zhao Chen, Eugene Lifshits, Anthony C. Faber, Alan Yeo, and Aaron N. Hata
- Abstract
PDF file - 11671KB, Supplemental Figure 1. Response of Kras mouse models to MEKi/PI3Ki. Supplemental Figure 2. MEKi/PI3Ki induces G1 arrest and inhibits proliferation of KRAS mutant NSCLC cell lines. Supplemental Figure 3. Combined MEK and PI3K inhibition is necessary for maximal reduction in cell proliferation. Supplemental Figure 4. Apoptotic response of KRAS mutant NSCLC cell lines in response to MEKi/PI3Ki. Supplemental Figure 5. Mutational status of TP53 or STK11/LKB1 does not correlate with apoptotic response to MEKi/PI3Ki. Supplemental Figure 6. Secreted Gaussia luciferase allows for precise quantitation of tumor growth and treatment response. Supplemental Figure 7. Tumor response of KRAS NSCLC xenografts to MEKi/PI3Ki. Supplemental Figure 8. Apoptosis induced by MEKi/PI3Ki is BAX and caspase-3 dependent. Supplemental Figure 9. Modulation of MEK/ERK and PI3Ki/AKT transcriptional output does not correlate with apoptotic sensitivity of KRAS mutant NSCLC cancer cells. Supplemental Figure 10. Modulation of RalGDS signaling does not correlate with apoptotic sensitivity of KRAS mutant NSCLC cell lines. Supplemental Figure 11. Sensitivity to MEKi/PI3Ki does not correlate with BH3 priming. Supplemental Figure 12. siRNA mediated knockdown of PUMA and BIM protects from apoptosis induced by MEKi/PI3Ki. Supplemental Figure 13. Inhibition of BCL-2 alone does not restore apoptotic response in insensitive KRAS mutant NSCLC cell lines. Supplemental Figure 14. Individual protein expression levels of BCL-2 family members do not correlate with sensitivity to MEKi/PI3Ki. Supplemental Figure 15. Inducible ectopic expression of BIM. Supplemental Figure 16. Restoration of apoptosis by ABT-263 leads to MEKi/PI3Ki-induced regression in vivo. Supplemental Figure 17. In vitro derived MEKi/PI3Ki resistant cells. Supplemental Figure 18. Cell lines derived from resistant Kras p53L/L tumors. Supplemental Figure 19. PUMA and BIM mRNA levels do not differ between cell lines derived from treatment naive and resistant Kras p53L/L tumors. Supplemental Table 1. Mutational status of KRAS mutant NSCLC cell lines. Supplemental Table 2. Knockdown efficiency of pLKO shRNA hairpins used in lentiviral screen.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Table 1 from PF00299804, an Irreversible Pan-ERBB Inhibitor, Is Effective in Lung Cancer Models with EGFR and ERBB2 Mutations that Are Resistant to Gefitinib
- Author
-
Pasi A. Jänne, Kwok-Kin Wong, Matthew Meyerson, John V. Heymach, James M. Nelson, Geoffrey I. Shapiro, Leena Gandhi, Irene W. Althaus, James E. Bradner, George N. Naumov, Patrick W. Vincent, Feng Zhao, Takeshi Shimamura, Andrea J. Gonzales, Eugene Lifshits, Christopher-Michael Gale, Kreshnik Zejnullahu, and Jeffrey A. Engelman
- Abstract
Supplementary Table 1 from PF00299804, an Irreversible Pan-ERBB Inhibitor, Is Effective in Lung Cancer Models with EGFR and ERBB2 Mutations that Are Resistant to Gefitinib
- Published
- 2023
- Full Text
- View/download PDF
9. Data from PF00299804, an Irreversible Pan-ERBB Inhibitor, Is Effective in Lung Cancer Models with EGFR and ERBB2 Mutations that Are Resistant to Gefitinib
- Author
-
Pasi A. Jänne, Kwok-Kin Wong, Matthew Meyerson, John V. Heymach, James M. Nelson, Geoffrey I. Shapiro, Leena Gandhi, Irene W. Althaus, James E. Bradner, George N. Naumov, Patrick W. Vincent, Feng Zhao, Takeshi Shimamura, Andrea J. Gonzales, Eugene Lifshits, Christopher-Michael Gale, Kreshnik Zejnullahu, and Jeffrey A. Engelman
- Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors gefitinib and erlotinib are effective treatments for a subset of non–small cell lung cancers. In particular, cancers with specific EGFR-activating mutations seem to be the most sensitive to these agents. However, despite their initial response, such cancers almost invariably develop resistance. In 50% of such cancers, a secondary EGFR mutation, T790M, has been identified that renders gefitinib and erlotinib ineffective inhibitors of EGFR kinase activity. Thus, there is a clinical need to develop novel EGFR inhibitors that can effectively inactivate T790M-containing EGFR proteins. In this study, we evaluate the effectiveness of a novel compound, PF00299804, an irreversible pan-ERBB inhibitor. The results from these studies show that PF00299804 is a potent inhibitor of EGFR-activating mutations as well as the EGFR T790M resistance mutation both in vitro and in vivo. Additionally, PF00299804 is a highly effective inhibitor of both the wild-type ERBB2 and the gefitinib-resistant oncogenic ERBB2 mutation identified in lung cancers. These preclinical evaluations support further clinical development of PF00299804 for cancers with mutations and/or amplifications of ERBB family members. [Cancer Res 2007;67(24):11924–32]
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figures 1-11, Methods from Multiple Mutations and Bypass Mechanisms Can Contribute to Development of Acquired Resistance to MET Inhibitors
- Author
-
Jeffrey A. Engelman, Pasi A. Jänne, James G. Christensen, Eugene Lifshits, Andrew Rogers, Michele A. McTigue, and Jie Qi
- Abstract
Supplementary Figures 1-11, Methods from Multiple Mutations and Bypass Mechanisms Can Contribute to Development of Acquired Resistance to MET Inhibitors
- Published
- 2023
- Full Text
- View/download PDF
11. PF-06463922, an ALK/ROS1 Inhibitor, Overcomes Resistance to First and Second Generation ALK Inhibitors in Preclinical Models
- Author
-
Wenyue Hu, Rosa L. Frias, Hovhannes J. Gukasyan, Alice T. Shaw, Valeria Fantin, Ryohei Katayama, Nathan V. Lee, Ruth W. Tang, Timothy Affolter, Eugene Lifshits, Ted William Johnson, Divya Bezwada, David P. Kodack, Lars D. Engstrom, Hieu Lam, Sidra Mahmood, Tod Smeal, Luc Friboulet, Rakesh K. Jain, Hui Wang, Melissa West, Dac M. Dinh, Bhushankumar Patel, Qiuhua Li, Konstantinos Tsaparikos, Helen Y. Zou, Justine L. Lam, Sergei Timofeevski, Shinji Yamazaki, Patrick B. Lappin, Justin F. Gainor, Shibing Deng, and Jinwei Wang
- Subjects
Alectinib ,Cancer Research ,Mutation ,Brigatinib ,Ceritinib ,business.industry ,Cell Biology ,Drug resistance ,Pharmacology ,medicine.disease_cause ,Lorlatinib ,3. Good health ,Oncology ,hemic and lymphatic diseases ,medicine ,ROS1 ,Anaplastic lymphoma kinase ,business ,medicine.drug - Abstract
SummaryWe report the preclinical evaluation of PF-06463922, a potent and brain-penetrant ALK/ROS1 inhibitor. Compared with other clinically available ALK inhibitors, PF-06463922 displayed superior potency against all known clinically acquired ALK mutations, including the highly resistant G1202R mutant. Furthermore, PF-06463922 treatment led to regression of EML4-ALK-driven brain metastases, leading to prolonged mouse survival, in a superior manner. Finally, PF-06463922 demonstrated high selectivity and safety margins in a variety of preclinical studies. These results suggest that PF-06463922 will be highly effective for the treatment of patients with ALK-driven lung cancers, including those who relapsed on clinically available ALK inhibitors because of secondary ALK kinase domain mutations and/or brain metastases.
- Published
- 2015
- Full Text
- View/download PDF
12. Patient-derived models of acquired resistance can identify effective drug combinations for cancer
- Author
-
Ryohei Katayama, Patricia Greninger, Justin F. Gainor, Anuj Kalsy, Leila Elamine, Sridhar Ramaswamy, Cyril H. Benes, Eugene Lifshits, Mari Mino-Kenudson, Maria Gomez-Caraballo, Jeffrey A. Engelman, Hayley Robinson, Dana Lee, Elizabeth L. Lockerman, Anthony C. Faber, Wooyoung Hur, Alice T. Shaw, Matthew J. Niederst, Lecia V. Sequist, Arnaud Amzallag, Emily Howe, Adam S. Crystal, A. John Iafrate, Mark M. Awad, Luc Friboulet, and Rosa L. Frias
- Subjects
Patient-Specific Modeling ,Lung Neoplasms ,DNA Mutational Analysis ,Proto-Oncogene Proteins pp60(c-src) ,MAP Kinase Kinase 1 ,Drug resistance ,Carcinoma, Non-Small-Cell Lung ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Cells, Cultured ,Humans ,Receptor, Fibroblast Growth Factor, Type 3 ,Anaplastic lymphoma kinase ,Medicine ,Anaplastic Lymphoma Kinase ,Molecular Targeted Therapy ,Sulfones ,Epidermal growth factor receptor ,Lung cancer ,Protein Kinase Inhibitors ,Multidisciplinary ,biology ,business.industry ,Receptor Protein-Tyrosine Kinases ,Cancer ,medicine.disease ,Enzyme Activation ,ErbB Receptors ,Pyrimidines ,Drug Resistance, Neoplasm ,Fibroblast growth factor receptor ,Mutation ,Cancer research ,biology.protein ,Drug Screening Assays, Antitumor ,business ,Tyrosine kinase ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Targeted cancer therapies have produced substantial clinical responses, but most tumors develop resistance to these drugs. Here, we describe a pharmacogenomic platform that facilitates rapid discovery of drug combinations that can overcome resistance. We established cell culture models derived from biopsy samples of lung cancer patients whose disease had progressed while on treatment with epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors and then subjected these cells to genetic analyses and a pharmacological screen. Multiple effective drug combinations were identified. For example, the combination of ALK and MAPK kinase (MEK) inhibitors was active in an ALK -positive resistant tumor that had developed a MAP2K1 activating mutation, and the combination of EGFR and fibroblast growth factor receptor (FGFR) inhibitors was active in an EGFR mutant resistant cancer with a mutation in FGFR3 . Combined ALK and SRC (pp60c-src) inhibition was effective in several ALK-driven patient-derived models, a result not predicted by genetic analysis alone. With further refinements, this strategy could help direct therapeutic choices for individual patients.
- Published
- 2014
- Full Text
- View/download PDF
13. Spontaneous Reversion of the Angiogenic Phenotype to a Nonangiogenic and Dormant State in Human Tumors
- Author
-
Michael Lampa, Randolph S. Watnick, Oddbjørn Straume, Trond Hellem Bø, Katherine Novak, Michael S. Rogers, Lars A. Akslen, George N. Naumov, Lorna M. Cryan, Eugene Lifshits, Elise Bender, Kamila Naxerova, Judah Folkman, Anna Blois, Karl-Henning Kalland, Soo-Young Kang, Anne Margrete Øyan, and David Zurakowski
- Subjects
Male ,Cancer Research ,Angiogenic Switch ,Population ,Clone (cell biology) ,Reversion ,Gene Expression ,Cell Growth Processes ,Mice, SCID ,Biology ,Article ,Mice ,In vivo ,Cell Line, Tumor ,Animals ,Humans ,education ,Molecular Biology ,education.field_of_study ,Neovascularization, Pathologic ,Liposarcoma ,Phenotype ,Oncology ,Tumor progression ,Cell culture ,Cancer research ,Heterografts - Abstract
The angiogenic switch, a rate-limiting step in tumor progression, has already occurred by the time most human tumors are detectable. However, despite significant study of the mechanisms controlling this switch, the kinetics and reversibility of the process have not been explored. The stability of the angiogenic phenotype was examined using an established human liposarcoma xenograft model. Nonangiogenic cells inoculated into immunocompromised mice formed microscopic tumors that remained dormant for approximately 125 days (vs. 95%) initiated angiogenic growth with second-order kinetics. These original, clonally derived angiogenic tumor cells were passaged through four in vivo cycles. At each cycle, a new set of single-cell clones was established from the most angiogenic clone and characterized for in vivo for tumorigenic activity. A total of 132 single-cell clones were tested in the second, third, and fourth in vivo passage. Strikingly, at each passage, a portion of the single-cell clones formed microscopic, dormant tumors. Following dormancy, like the original cell line, these revertant tumors spontaneously switched to the angiogenic phenotype. Finally, revertant clones were transcriptionally profiled and their angiogenic output determined. Collectively, these data demonstrate that the angiogenic phenotype in tumors is malleable and can spontaneously revert to the nonangiogenic phenotype in a population of human tumor cells. Implications: Leveraging the rate of reversion to the nonangiogenic phenotype and tumor dormancy may be a novel anticancer strategy. Mol Cancer Res; 12(5); 754–64. ©2014 AACR.
- Published
- 2014
- Full Text
- View/download PDF
14. Receptor tyrosine kinases exert dominant control over PI3K signaling in human KRAS mutant colorectal cancers
- Author
-
David P. Ryan, Kwok-Kin Wong, Lewis C. Cantley, Ryan B. Corcoran, Youngchul Song, Cyril H. Benes, Hiromichi Ebi, Jeffrey A. Meyerhardt, Jeffrey A. Engelman, Anurag K. Singh, Zhao Chen, Eugene Lifshits, and Jeffrey Settleman
- Subjects
Male ,Proto-Oncogene Proteins B-raf ,MAPK/ERK pathway ,Class I Phosphatidylinositol 3-Kinases ,MAP Kinase Signaling System ,Transplantation, Heterologous ,Mice, Nude ,Apoptosis ,Biology ,medicine.disease_cause ,Receptor tyrosine kinase ,Proto-Oncogene Proteins p21(ras) ,Mice ,Phosphatidylinositol 3-Kinases ,Downregulation and upregulation ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,RNA, Small Interfering ,Protein Kinase Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,DNA Primers ,Base Sequence ,Receptor Protein-Tyrosine Kinases ,General Medicine ,Transplantation ,Gene Knockdown Techniques ,Mutation ,ras Proteins ,Cancer research ,biology.protein ,KRAS ,Signal transduction ,Colorectal Neoplasms ,Proto-Oncogene Proteins c-akt ,Neoplasm Transplantation ,Signal Transduction ,Research Article - Abstract
Therapies inhibiting receptor tyrosine kinases (RTKs) are effective against some human cancers when they lead to simultaneous downregulation of PI3K/AKT and MEK/ERK signaling. However, mutant KRAS has the capacity to directly activate ERK and PI3K signaling, and this is thought to underlie the resistance of KRAS mutant cancers to RTK inhibitors. Here, we have elucidated the molecular regulation of both the PI3K/AKT and MEK/ERK signaling pathways in KRAS mutant colorectal cancer cells and identified combination therapies that lead to robust cancer cell apoptosis. KRAS knockdown using shRNA suppressed ERK signaling in all of the human KRAS mutant colorectal cancer cell lines examined. However, no decrease, and actually a modest increase, in AKT phosphorylation was often seen. By performing PI3K immunoprecipitations, we determined that RTKs, often IGF-IR, regulated PI3K signaling in the KRAS mutant cell lines. This conclusion was also supported by the observation that specific RTK inhibition led to marked suppression of PI3K signaling and biochemical assessment of patient specimens. Interestingly, combination of RTK and MEK inhibitors led to concomitant inhibition of PI3K and MEK signaling, marked growth suppression, and robust apoptosis of human KRAS mutant colorectal cancer cell lines in vitro and upon xenografting in mice. These findings provide a framework for utilizing RTK inhibitors in the treatment of KRAS mutant colorectal cancers.
- Published
- 2011
- Full Text
- View/download PDF
15. Therapeutic strategies to overcome crizotinib resistance in non-small cell lung cancers harboring the fusion oncogene EML4-ALK
- Author
-
Victor M. Rivera, Ryohei Katayama, Alice T. Shaw, Jeffrey A. Engelman, William C Shakespeare, Cyril H. Benes, Eugene Lifshits, Anthony J. Iafrate, Tahsin M. Khan, and Hiromichi Ebi
- Subjects
EML4/ALK Fusion Gene ,Oncogene Proteins, Fusion ,Pyridines ,medicine.drug_class ,Immunoblotting ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Drug resistance ,Biology ,Transfection ,Tyrosine-kinase inhibitor ,Hsp90 inhibitor ,Mice ,Organophosphorus Compounds ,Crizotinib ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Phosphorylation ,RNA, Small Interfering ,In Situ Hybridization, Fluorescence ,DNA Primers ,Multidisciplinary ,Dose-Response Relationship, Drug ,Oncogene ,Ceritinib ,Reverse Transcriptase Polymerase Chain Reaction ,Biological Sciences ,Flow Cytometry ,Survival Analysis ,ALK inhibitor ,Pyrimidines ,Drug Resistance, Neoplasm ,Mutation ,Immunology ,Cancer research ,Pyrazoles ,Signal Transduction ,medicine.drug - Abstract
The echinoderm microtubule-associated protein-like 4 (EML4)-anaplastic lymphoma kinase (ALK) fusion oncogene represents a molecular target in a small subset of non-small cell lung cancers (NSCLCs). This fusion leads to constitutive ALK activation with potent transforming activity. In a pivotal phase 1 clinical trial, the ALK tyrosine kinase inhibitor (TKI) crizotinib (PF-02341066) demonstrated impressive antitumor activity in the majority of patients with NSCLC harboring ALK fusions. However, despite these remarkable initial responses, cancers eventually develop resistance to crizotinib, usually within 1 y, thereby limiting the potential clinical benefit. To determine how cancers acquire resistance to ALK inhibitors, we established a model of acquired resistance to crizotinib by exposing a highly sensitive EML4-ALK–positive NSCLC cell line to increasing doses of crizotinib until resistance emerged. We found that cells resistant to intermediate doses of crizotinib developed amplification of the EML4-ALK gene. Cells resistant to higher doses (1 μM) also developed a gatekeeper mutation, L1196M, within the kinase domain, rendering EML4-ALK insensitive to crizotinib. This gatekeeper mutation was readily detected using a unique and highly sensitive allele-specific PCR assay. Although crizotinib was ineffectual against EML4-ALK harboring the gatekeeper mutation, we observed that two structurally different ALK inhibitors, NVP-TAE684 and AP26113, were highly active against the resistant cancer cells in vitro and in vivo. Furthermore, these resistant cells remained highly sensitive to the Hsp90 inhibitor 17-AAG. Thus, we have developed a model of acquired resistance to ALK inhibitors and have shown that second-generation ALK TKIs or Hsp90 inhibitors are effective in treating crizotinib-resistant tumors harboring secondary gatekeeper mutations.
- Published
- 2011
- Full Text
- View/download PDF
16. Multiple Mutations and Bypass Mechanisms Can Contribute to Development of Acquired Resistance to MET Inhibitors
- Author
-
Michele McTigue, Andrew H. Rogers, Jie Qi, Pasi A. Jänne, Jeffrey A. Engelman, Eugene Lifshits, and James G. Christensen
- Subjects
Cancer Research ,TGF alpha ,Indoles ,Pyridines ,Mice, Nude ,Drug resistance ,medicine.disease_cause ,Article ,Receptor tyrosine kinase ,Mice ,Crizotinib ,Stomach Neoplasms ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Receptors, Growth Factor ,Sulfones ,Epidermal growth factor receptor ,Extracellular Signal-Regulated MAP Kinases ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Mutation ,biology ,MAP kinase kinase kinase ,Proto-Oncogene Proteins c-met ,Transforming Growth Factor alpha ,MAP Kinase Kinase Kinases ,Xenograft Model Antitumor Assays ,Elafin ,ErbB Receptors ,Oncogene Protein v-akt ,Oncology ,Drug Resistance, Neoplasm ,Immunology ,Cancer research ,biology.protein ,Pyrazoles ,Transcription Factors ,Transforming growth factor - Abstract
Therapies targeting receptor tyrosine kinases have shown efficacy in molecularly defined subsets of cancers. Unfortunately, cancers invariably develop resistance, and overcoming or preventing resistance will ultimately be key to unleashing their full therapeutic potential. In this study, we examined how cancers become resistant to MET inhibitors, a class of drugs currently under clinical development. We utilized the highly sensitive gastric carcinoma cell line, SNU638, and two related MET inhibitors PHA-665752 and PF-2341066. To our surprise, we observed at least two mechanisms of resistance that arose simultaneously. Both resulted in maintenance of downstream PI3K (phosphoinositide 3-kinase)-AKT and MEK (MAP/ERK kinase)-ERK signaling in the presence of inhibitor. One mechanism, observed by modeling resistance both in vitro and in vivo, involved the acquisition of a mutation in the MET activation loop (Y1230). Structural analysis indicates that this mutation destabilizes the autoinhibitory conformation of MET and abrogates an important aromatic stacking interaction with the inhibitor. The other cause of resistance was activation of the epidermal growth factor receptor (EGFR) pathway due to increased expression of transforming growth factor α. Activation of EGFR bypassed the need for MET signaling to activate downstream signaling in these cells. This resistance could be overcome by combined EGFR and MET inhibition. Thus, therapeutic strategies that combine MET inhibitors capable of inhibiting Y1230 mutant MET in combination with anti-EGFR–based therapies may enhance clinical benefit for patients with MET-addicted cancers. Importantly, these results also underscore the notion that a single cancer can simultaneously develop resistance induced by several mechanisms and highlight the daunting challenges associated with preventing or overcoming resistance. Cancer Res; 71(3); 1081–91. ©2011 AACR.
- Published
- 2011
- Full Text
- View/download PDF
17. Differential induction of apoptosis in HER2 and EGFR addicted cancers following PI3K inhibition
- Author
-
Roderick T. Bronson, Beow Y. Yeap, Danan Li, Kwok-Kin Wong, Carlos Garcia-Echeverria, Eugene Lifshits, Young Chul Song, Zhao Chen, Anthony C. Faber, Mei-Chih Liang, Jeffrey A. Engelman, and Sauveur Michel Maira
- Subjects
Lung Neoplasms ,Receptor, ErbB-2 ,Apoptosis ,Breast Neoplasms ,Biology ,Erlotinib Hydrochloride ,Mice ,Gefitinib ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Epidermal growth factor receptor ,Protein Kinase Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Multidisciplinary ,TOR Serine-Threonine Kinases ,Imidazoles ,Biological Sciences ,MAP Kinase Kinase Kinases ,Xenograft Model Antitumor Assays ,respiratory tract diseases ,ErbB Receptors ,Quinazolines ,Quinolines ,Cancer research ,biology.protein ,Female ,Erlotinib ,Signal transduction ,Protein Kinases ,medicine.drug - Abstract
Non-small cell lung cancers with activating mutations in the epidermal growth factor receptor (EGFR) are highly responsive to EGFR tyrosine kinase inhibitors (TKIs), such as gefitinib and erlotinib. Such cancers are “addicted” to EGFR, and treatment with a TKI invariably leads to down-regulation of the PI3K-AKT-mTOR and MEK-ERK signaling pathways, resulting in apoptosis. Using a dual PI3K-mTOR inhibitor, NVP-BEZ235, we evaluated whether PI3K-mTOR inhibition alone induced apoptosis in these cancers. In contrast toHER2-amplified breast cancers, we found that PI3K-mTOR inhibition did not promote substantial apoptosis in theEGFRmutant lung cancers. However, blocking both PI3K-mTOR and MEK simultaneously led to apoptosis to similar levels as the EGFR TKIs, suggesting that down-regulation of these pathways may account for much of the apoptosis promoted by EGFR inhibition. InEGFRmutant lung cancers, down-regulation of both intracellular pathways converged on the BH3 family of proteins regulating apoptosis. PI3K inhibition led to down-regulation of Mcl-1, and MEK inhibition led to up-regulation of BIM. In fact, down-regulation of Mcl-1 by siRNA was sufficient to sensitize these cancers to single-agent MEK inhibitors. Surprisingly, an AKT inhibitor did not decrease Mcl-1 levels, and when combined with MEK inhibitors, failed to induce apoptosis. Importantly, we observed that the combination of PI3K-mTOR and MEK inhibitors effectively shrunk tumors in a transgenic and xenograft model ofEGFRT790M-L858R cancers. These data indicate simultaneous inhibition of PI3K-mTOR and MEK signaling is an effective strategy for treatingEGFRmutant lung cancers, including those with acquired resistance to EGFR TKIs.
- Published
- 2009
- Full Text
- View/download PDF
18. Combined Vascular Endothelial Growth Factor Receptor and Epidermal Growth Factor Receptor (EGFR) Blockade Inhibits Tumor Growth in Xenograft Models of EGFR Inhibitor Resistance
- Author
-
John V. Heymach, Eugene Lifshits, George N. Naumov, Tina Cascone, Daniel G. Tenen, Bruce E. Johnson, Lars A. Akslen, Jeffrey A. Engelman, Monique B. Nilsson, Hua Kang Wu, Oddbjørn Straume, Pasi A. Jänne, Beow Y. Yeap, Balazs Halmos, Xi M. Tang, Alexandra Briggs, Judah Folkman, Lauren Averett Byers, Li Xu, and Susumu Kobayashi
- Subjects
Vascular Endothelial Growth Factor A ,Cancer Research ,Lung Neoplasms ,Time Factors ,Blotting, Western ,Mice, Nude ,Biology ,Antibodies, Monoclonal, Humanized ,Transfection ,medicine.disease_cause ,Vandetanib ,Article ,Erlotinib Hydrochloride ,Mice ,Gefitinib ,Piperidines ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Epidermal growth factor receptor ,neoplasms ,Cell Proliferation ,EGFR inhibitors ,Reverse Transcriptase Polymerase Chain Reaction ,Antibodies, Monoclonal ,Xenograft Model Antitumor Assays ,Tumor Burden ,respiratory tract diseases ,Bevacizumab ,ErbB Receptors ,Receptors, Vascular Endothelial Growth Factor ,Oncology ,Drug Resistance, Neoplasm ,Mutation ,NIH 3T3 Cells ,Quinazolines ,Cancer research ,biology.protein ,KRAS ,Erlotinib ,Tyrosine kinase ,medicine.drug - Abstract
Purpose: The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI) gefitinib and erlotinib benefit some non–small cell lung cancer (NSCLC) patients, but most do not respond (primary resistance) and those who initially respond eventually progress (acquired resistance). EGFR TKI resistance is not completely understood and has been associated with certain EGFR and K-RAS mutations and MET amplification.Experimental Design: We hypothesized that dual inhibition of the vascular endothelial growth factor (VEGF) and EGFR pathways may overcome primary and acquired resistance. We investigated the VEGF receptor/EGFR TKI vandetanib, and the combination of bevacizumab and erlotinib in vivo using xenograft models of EGFR TKI sensitivity, primary resistance, and three models of acquired resistance, including models with mutated K-RAS and secondary EGFR T790M mutation.Results: Vandetanib, gefitinib, and erlotinib had similar profiles of in vitro activity and caused sustained tumor regressions in vivo in the sensitive HCC827 model. In all four resistant models, vandetanib and bevacizumab/erlotinib were significantly more effective than erlotinib or gefitinib alone. Erlotinib resistance was associated with a rise in both host and tumor-derived VEGF but not EGFR secondary mutations in the KRAS mutant-bearing A549 xenografts. Dual inhibition reduced tumor endothelial proliferation compared with VEGF or EGFR blockade alone, suggesting that the enhanced activity of dual inhibition is due at least in part to antiendothelial effects.Conclusion: These studies suggest that erlotinib resistance may be associated with a rise in both tumor cell and host stromal VEGF and that combined blockade of the VEGFR and EGFR pathways can abrogate primary or acquired resistance to EGFR TKIs. This approach merits further evaluation in NSCLC patients.
- Published
- 2009
- Full Text
- View/download PDF
19. Targeting EGFR activity in blood vessels is sufficient to inhibit tumor growth and is accompanied by an increase in VEGFR-2 dependence in tumor endothelial cells
- Author
-
Diane R. Bielenberg, John V. Heymach, Michael Klagsbrun, Eugene Lifshits, and Dhara N. Amin
- Subjects
Male ,Receptor, ErbB-2 ,Angiogenesis ,Transplantation, Heterologous ,Mice, Nude ,Biochemistry ,Cell Line ,Mice ,Gefitinib ,Cell Line, Tumor ,medicine ,Animals ,Humans ,ERBB3 ,Epidermal growth factor receptor ,Enzyme Inhibitors ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase B ,Cell Proliferation ,Epidermal Growth Factor ,biology ,Chemistry ,Kinase ,Melanoma ,Endothelial Cells ,Neoplasms, Experimental ,Cell Biology ,Tyrphostins ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-2 ,Neuropilin-1 ,ErbB Receptors ,Cinnamates ,Quinazolines ,Cancer research ,biology.protein ,Cyclin-dependent kinase 8 ,Endothelium, Vascular ,Cardiology and Cardiovascular Medicine ,Proto-Oncogene Proteins c-akt ,Signal Transduction ,medicine.drug - Abstract
Epidermal growth factor receptor (EGFR) targeting agents such as kinase inhibitors reduce tumor growth and progression. We have previously reported that EGFR is not only expressed by the tumor cells but by the tumor endothelial cells (EC) as well (Amin, D. N., Hida, K., Bielenberg, D. R., Klagsbrun, M., 2006. Tumor endothelial cells express epidermal growth factor receptor (EGFR) but not ErbB3 and are responsive to EGF and to EGFR kinase inhibitors. Cancer Res. 66, 2173-80). Thus, targeting tumor blood vessel EGFR may be a viable strategy for tumor growth inhibition. We describe here a melanoma xenograft model where the tumor cells express very little or no EGFR but the tumor blood vessels express activated EGFR. The EGFR kinase inhibitor, gefitinib (Iressa), retarded tumor growth with a size decrease of 38% compared to control mice, ostensibly due to targeting of the blood vessels. EC were isolated from tumors of gefitinib-treated mice. These EC were unable to proliferate in response to EGF and displayed relatively weaker activation of MAPK and AKT signaling in response to EGF compared to tumor EC isolated from vehicle-treated mice. In contrast, the tumor EC from gefitinib-treated mice expressed higher levels of VEGFR-2 both at the mRNA and protein level. In addition, these cells were less sensitive to EGFR kinase inhibitors in vitro but more sensitive to a VEGFR-2 kinase inhibitor. These results suggest that in tumor EC from gefitinib-treated mice there is a switch from dependence on EGFR activity to signaling via VEGFR-2. Our data provide a molecular rationale for combination therapies targeting both EGF and VEGF signaling on the tumor vasculature.
- Published
- 2008
- Full Text
- View/download PDF
20. Allelic dilution obscures detection of a biologically significant resistance mutation in EGFR-amplified lung cancer
- Author
-
Lewis C. Cantley, George N. Naumov, John V. Heymach, Ana M. Borras, Bruce E. Johnson, Christopher Michael Gale, Toru Mukohara, Emily T Jarrell, Beow Y. Yeap, Pasi A. Jänne, Jeffrey A. Engelman, Jason Sun, Kreshnik Zejnullahu, Sean Tracy, Xiaojun Zhao, and Eugene Lifshits
- Subjects
Lung Neoplasms ,Receptor, ErbB-3 ,Mutant ,Mutation, Missense ,Apoptosis ,Biology ,Transfection ,medicine.disease_cause ,Mice ,Phosphatidylinositol 3-Kinases ,T790M ,Gefitinib ,Cell Line, Tumor ,medicine ,Animals ,Humans ,heterocyclic compounds ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,skin and connective tissue diseases ,Lung cancer ,Protein Kinase Inhibitors ,neoplasms ,Alleles ,Cell Proliferation ,EGFR inhibitors ,Mutation ,Base Sequence ,Gene Amplification ,General Medicine ,medicine.disease ,Resistance mutation ,Xenograft Model Antitumor Assays ,Molecular biology ,respiratory tract diseases ,ErbB Receptors ,Drug Resistance, Neoplasm ,Quinazolines ,Cancer research ,RNA Interference ,Erlotinib ,Proto-Oncogene Proteins c-akt ,Research Article ,medicine.drug - Abstract
EGFR is frequently mutated and amplified in lung adenocarcinomas sensitive to EGFR inhibitors gefitinib and erlotinib. A secondary mutation, T790M, has been associated with acquired resistance but has not been shown to be sufficient to render EGFR mutant/amplified lung cancers resistant to EGFR inhibitors. We created a model for studying acquired resistance to gefitinib by prolonged exposure of a gefitinib-sensitive lung carcinoma cell line (H3255; EGFR mutated and amplified) to gefitinib in vitro. The resulting resistant cell line acquired a T790M mutation in a small fraction of the amplified alleles that was undetected by direct sequencing and identified only by a highly sensitive HPLC-based technique. In gefitinib-sensitive lung cancer cells with EGFR mutations and amplifications, exogenous introduction of EGFR T790M effectively conferred resistance to gefitinib and continued ErbB-3/PI3K/Akt signaling when in cis to an activating mutation. Moreover, continued activation of PI3K signaling by the PIK3CA oncogenic mutant, p110alpha E545K, was sufficient to abrogate gefitinib-induced apoptosis. These findings suggest that allelic dilution of biologically significant resistance mutations may go undetected by direct sequencing in cancers with amplified oncogenes and that restoration of PI3K activation via either a T790M mutation or other mechanisms can provide resistance to gefitinib.
- Published
- 2006
- Full Text
- View/download PDF
21. Failure to induce apoptosis via BCL-2 family proteins underlies lack of efficacy of combined MEK and PI3K inhibitors for KRAS mutant lung cancers
- Author
-
Eugene Lifshits, Anthony C. Faber, Zhao Chen, Katherine A. Cheng, Kwok-Kin Wong, Alan Yeo, Anthony Letai, Aaron N. Hata, Zandra E. Walton, Kristopher A. Sarosiek, Rebecca S. Heist, Jeffrey A. Engelman, and Mari Mino-Kenudson
- Subjects
Cancer Research ,Lung Neoplasms ,MAP Kinase Signaling System ,bcl-X Protein ,Mice, Nude ,Apoptosis ,medicine.disease_cause ,Article ,Small hairpin RNA ,Mice ,Phosphatidylinositol 3-Kinases ,In vivo ,Puma ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Animals ,Humans ,neoplasms ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,biology ,Bcl-2 family ,biology.organism_classification ,Cell biology ,respiratory tract diseases ,Mitochondria ,Oncology ,Proto-Oncogene Proteins c-bcl-2 ,Mutation ,ras Proteins ,KRAS ,Signal transduction ,Apoptosis Regulatory Proteins ,Signal Transduction - Abstract
Although several groups have demonstrated that concomitant use of MEK and phosphoinositide 3-kinase (PI3K) inhibitors (MEKi/PI3Ki) can induce dramatic tumor regressions in mouse models of KRAS-mutant non–small cell lung cancer (NSCLC), ongoing clinical trials investigating this strategy have been underwhelming to date. While efficacy may be hampered by a narrow therapeutic index, the contribution of biologic heterogeneity in the response of KRAS-mutant NSCLCs to MEKi/PI3Ki has been largely unexplored. In this study, we find that most human KRAS-mutant NSCLC cell lines fail to undergo marked apoptosis in response to MEKi/PI3Ki, which is key for tumor responsiveness in vivo. This heterogeneity of apoptotic response occurs despite relatively uniform induction of growth arrest. Using a targeted short hairpin RNA screen of BCL-2 family members, we identify BIM, PUMA, and BCL-XL as key regulators of the apoptotic response induced by MEKi/PI3Ki, with decreased expression of BIM and PUMA relative to BCL-XL in cell lines with intrinsic resistance. In addition, by modeling adaptive resistance to MEKi/PI3Ki both in vitro and in vivo, we find that, upon the development of resistance, tumors have a diminished apoptotic response due to downregulation of BIM and PUMA. These results suggest that the inability to induce apoptosis may limit the effectiveness of MEKi/PI3Ki for KRAS-mutant NSCLCs by contributing to intrinsic and adaptive resistance to this therapy. Cancer Res; 74(11); 3146–56. ©2014 AACR.
- Published
- 2014
22. BIM expression in treatment-naive cancers predicts responsiveness to kinase inhibitors
- Author
-
Henry L. Gomez, Miguel Rivera, Ryan B. Corcoran, Erik J. Coffman, Anthony C. Faber, Cyril H. Benes, Carlos L. Arteaga, Joao Incio, Dora Dias-Santagata, Belinda A. Waltman, Eugene Lifshits, Sylvie Roberge, Sarah F. Pollack, Jeffrey A. Engelman, Youngchul Song, Hiromichi Ebi, Lecia V. Sequist, Alona Muzikansky, Euiheon Chung, José Baselga, Subba R. Digumarthy, and Rakesh K. Jain
- Subjects
Lung Neoplasms ,Receptor, ErbB-2 ,Mutant ,Mice, Nude ,Apoptosis ,Biology ,Article ,Cohort Studies ,Mice ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Lung cancer ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,EGFR inhibitors ,Phosphoinositide-3 Kinase Inhibitors ,Retrospective Studies ,Oncogene ,Bcl-2-Like Protein 11 ,Kinase ,Membrane Proteins ,Oncogenes ,medicine.disease ,ErbB Receptors ,Oncology ,Cancer research ,Female ,Signal transduction ,Apoptosis Regulatory Proteins ,BH3 Interacting Domain Death Agonist Protein ,Signal Transduction - Abstract
Cancers with specific genetic mutations are susceptible to selective kinase inhibitors. However, there is a wide spectrum of benefit among cancers harboring the same sensitizing genetic mutations. Herein, we measured apoptotic rates among cell lines sharing the same driver oncogene following treatment with the corresponding kinase inhibitor. There was a wide range of kinase inhibitor-induced apoptosis despite comparable inhibition of the target and associated downstream signaling pathways. Surprisingly, pretreatment RNA levels of the BH3-only pro-apoptotic BIM strongly predicted the capacity of EGFR, HER2, and PI3K inhibitors to induce apoptosis in EGFR-mutant, HER2-amplified, and PIK3CA-mutant cancers, respectively, but BIM levels did not predict responsiveness to standard chemotherapies. Furthermore, BIM RNA levels in EGFR-mutant lung cancer specimens predicted response and duration of clinical benefit from EGFR inhibitors. These findings suggest assessment of BIM levels in treatment-naïve tumor biopsies may indicate the degree of benefit from single-agent kinase inhibitors in multiple oncogene-addiction paradigms. Significance: In several oncogene-addiction paradigms, assessment of BIM RNA levels identifies those cancers that fail to have substantial apoptotic responses to kinase inhibitors. BIM RNA levels may be assessed in diagnostic cancer specimens to predict which patients will receive less benefit from single-agent kinase inhibitors. Cancer Discovery: 1(4); 352–65. ©2011 AACR. Read the Commentary on this article by Yoshida and Haura, p. 289 This article is highlighted in the In This Issue feature, p. 275
- Published
- 2011
23. Amplification of EGFR T790M causes resistance to an irreversible EGFR inhibitor
- Author
-
Kreshnik Zejnullahu, Dalia Ercan, Pasi A. Jänne, Alison Brown, David J. Kwiatkowski, Jeffrey A. Engelman, Eugene Lifshits, Charles Lee, Andrew H. Rogers, Marzia Capelletti, Yun Xiao, Kimio Yonesaka, and James G. Christensen
- Subjects
Cancer Research ,Lung Neoplasms ,medicine.drug_class ,Biology ,Tyrosine-kinase inhibitor ,Article ,T790M ,Gefitinib ,Growth factor receptor ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Epidermal growth factor receptor ,Phosphorylation ,Molecular Biology ,Protein Kinase Inhibitors ,EGFR inhibitors ,Quinazolinones ,Gene Amplification ,respiratory tract diseases ,ErbB Receptors ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,Quinazolines ,Erlotinib ,Tyrosine kinase ,medicine.drug - Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors, gefitinib and erlotinib, are effective therapies against mutant non-small cell lung cancers (NSCLCs). Treatment is limited by the development of resistance in part explained by the gain of a secondary EGFR mutation, T790M, at the gatekeeper residue. Irreversible EGFR inhibitors, including PF00299804, are effective in vitro and in vivo against EGFR mutant tumors that contain EGFR T790M and are currently under clinical development. In this study we generate models of resistance to PF00299804, using cell lines with EGFR T790M, and demonstrate that the PF00299804 resistant models develop focal amplification of EGFR that preferentially involves the T790M-containing allele. These PF00299804 resistant cell lines remain dependent on EGFR for growth as downregulation of EGFR by shRNA compromises their viability. We demonstrate that resistance to PF00299804 arises, at least in part, through selection of a pre-existing EGFR T790M amplified clone both in vitro and using a xenograft model in vivo. Our findings demonstrate that EGFR T790M is a common resistance mechanism to both reversible, and when amplified, the irreversible EGFR kinase inhibitors further emphasizing the need to develop more potent therapies against EGFR T790M. The findings can be used to guide studies of patient tumor specimens from ongoing clinical trials of irreversible EGFR kinase inhibitors.
- Published
- 2010
24. Class 1A PI3K regulates vessel integrity during development and tumorigenesis
- Author
-
Eugene Lifshits, Aya Matsui, Cyril H. Benes, Hak Soo Choi, John V. Frangioni, Tina L. Yuan, Ji Luo, and Lewis C. Cantley
- Subjects
Heterozygote ,Endothelium ,Angiogenesis ,Vascular permeability ,Biology ,medicine.disease_cause ,Neovascularization ,Capillary Permeability ,Mice ,Phosphatidylinositol 3-Kinases ,Neoplasms ,medicine ,Animals ,PI3K/AKT/mTOR pathway ,Tumor microenvironment ,Multidisciplinary ,Neovascularization, Pathologic ,Biological Sciences ,Embryonic stem cell ,Mice, Mutant Strains ,Protein Subunits ,medicine.anatomical_structure ,Phenotype ,Immunology ,Cancer research ,Blood Vessels ,Endothelium, Vascular ,medicine.symptom ,Carcinogenesis - Abstract
PI3K is important in the regulation of growth, proliferation, and survival of tumor cells. We show that class 1A PI3K is also critical in the tumor microenvironment by regulating the integrity of the tumor vasculature. Using Tie2Cre-mediated deletion of the PI3K regulatory subunits (p85alpha, p55alpha, p50alpha, and p85beta), we generated mice with endothelial cell-specific loss of class 1A PI3K. Complete loss of all subunits caused acute embryonic lethality at E11.5 due to hemorrhaging, whereas retention of a single p85alpha allele yielded viable mice that survived to adulthood. These heterozygous mice exhibited no vascular defects until challenged with a pathological insult, such as tumor cells or high levels of VEGF. Under these pathological conditions, heterozygous mice exhibited localized vascular abnormalities, including vessel leakage and the inability to maintain large vessels, which caused a deceleration of tumorigenesis. Furthermore, we show that a PI3K inhibitor can mimic the effects of class 1A PI3K loss, which suggests that targeting class 1A PI3K may be a promising therapy for blocking tumor angiogenesis.
- Published
- 2008
25. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer
- Author
-
Pasi A. Jänne, David J. Sugarbaker, Christopher T. Ducko, William G. Richards, Jinseon Lee, Jhingook Kim, Eugene Lifshits, Craig H. Mermel, J. Paul Marcoux, Derek Y. Chiang, Carly Murphy, Charles Lee, Matthew Meyerson, Alison J. Holmes, Neal I. Lindeman, Roman K. Thomas, Nathanael S. Gray, Kreshnik Zejnullahu, Hwan Geun Choi, Jeffrey A. Engelman, and Jussi Koivunen
- Subjects
Male ,Cancer Research ,EML4/ALK Fusion Gene ,Lung Neoplasms ,Oncogene Proteins, Fusion ,medicine.drug_class ,Receptor, ErbB-2 ,Cell ,Antineoplastic Agents ,Apoptosis ,Biology ,Article ,Gene Expression Regulation, Enzymologic ,hemic and lymphatic diseases ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Humans ,Epidermal growth factor receptor ,Enzyme Inhibitors ,Lung cancer ,Aged ,Kinase ,ALK Gene Rearrangement ,Cancer ,Middle Aged ,medicine.disease ,respiratory tract diseases ,ALK inhibitor ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Female - Abstract
Purpose: The EML4-ALK fusion gene has been detected in ∼7% of Japanese non-small cell lung cancers (NSCLC). We determined the frequency of EML4-ALK in Caucasian NSCLC and in NSCLC cell lines. We also determined whether TAE684, a specific ALK kinase inhibitor, would inhibit the growth of EML4-ALK-containing cell lines in vitro and in vivo.Experimental Design: We screened 305 primary NSCLC [both U.S. (n = 138) and Korean (n = 167) patients] and 83 NSCLC cell lines using reverse transcription-PCR and by exon array analyses. We evaluated the efficacy of TAE684 against NSCLC cell lines in vitro and in vivo.Results: We detected four different variants, including two novel variants, of EML4-ALK using reverse transcription-PCR in 8 of 305 tumors (3%) and 3 of 83 (3.6%) NSCLC cell lines. All EML4-ALK-containing tumors and cell lines were adenocarcinomas. EML4-ALK was detected more frequently in NSCLC patients who were never or light (Conclusions: EML4-ALK is found in the minority of NSCLC. ALK kinase inhibitors alone or in combination may nevertheless be clinically effective treatments for NSCLC patients whose tumors contain EML4-ALK.
- Published
- 2008
26. PF00299804, an irreversible pan-ERBB inhibitor, is effective in lung cancer models with EGFR and ERBB2 mutations that are resistant to gefitinib
- Author
-
Kreshnik Zejnullahu, Kwok-Kin Wong, Irene W. Althaus, Patrick Vincent, Pasi A. Jänne, Gonzales Andrea, John V. Heymach, Feng Zhao, James E. Bradner, Christopher Michael Gale, Geoffrey I. Shapiro, James M. Nelson, Leena Gandhi, Jeffrey A. Engelman, George N. Naumov, Matthew Meyerson, Eugene Lifshits, and Takeshi Shimamura
- Subjects
Cancer Research ,Lung Neoplasms ,medicine.drug_class ,Mice, Nude ,Antineoplastic Agents ,Tyrosine-kinase inhibitor ,chemistry.chemical_compound ,T790M ,Mice ,Gefitinib ,ErbB ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Epidermal growth factor receptor ,Cloning, Molecular ,neoplasms ,EGFR inhibitors ,Adaptor Proteins, Signal Transducing ,Quinazolinones ,biology ,Oncogene Proteins v-erbB ,Dacomitinib ,respiratory tract diseases ,ErbB Receptors ,Oncology ,chemistry ,Cancer research ,biology.protein ,Quinazolines ,Erlotinib ,Cell Division ,medicine.drug - Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors gefitinib and erlotinib are effective treatments for a subset of non–small cell lung cancers. In particular, cancers with specific EGFR-activating mutations seem to be the most sensitive to these agents. However, despite their initial response, such cancers almost invariably develop resistance. In 50% of such cancers, a secondary EGFR mutation, T790M, has been identified that renders gefitinib and erlotinib ineffective inhibitors of EGFR kinase activity. Thus, there is a clinical need to develop novel EGFR inhibitors that can effectively inactivate T790M-containing EGFR proteins. In this study, we evaluate the effectiveness of a novel compound, PF00299804, an irreversible pan-ERBB inhibitor. The results from these studies show that PF00299804 is a potent inhibitor of EGFR-activating mutations as well as the EGFR T790M resistance mutation both in vitro and in vivo. Additionally, PF00299804 is a highly effective inhibitor of both the wild-type ERBB2 and the gefitinib-resistant oncogenic ERBB2 mutation identified in lung cancers. These preclinical evaluations support further clinical development of PF00299804 for cancers with mutations and/or amplifications of ERBB family members. [Cancer Res 2007;67(24):11924–32]
- Published
- 2007
27. Abstract 130: PF-06463922, a novel next generation ALK/ROS1 inhibitor, overcomes resistance to 1st and 2nd generation ALK inhibitors in pre-clinical models
- Author
-
Luc Friboulet, Tod Smeal, Konstantinos Tsaparikos, Jinwei Wang, Rakesh K. Jain, Hovhannes J. Gukasyan, Valeria Fantin, Qiuhua Li, Helen Y. Zou, Nathan V. Lee, Timothy Affolter, Melissa West, Patrick B. Lappin, Lars D. Engstrom, Sergei Timofeevski, Ted William Johnson, Eugene Lifshits, Justine L. Lam, Hui Wang, Dac M. Dinh, Bhushankumar Patel, David P. Kodack, Hieu Lam, Sidra Mahmood, Shibing Deng, Shinji Yamazaki, Divya Bezwada, Alice T. Shaw, Wenyue Hu, and Ruth W. Tang
- Subjects
Gerontology ,Cancer Research ,Kinase ,business.industry ,Cancer ,Drug resistance ,medicine.disease ,Oncology ,Protein kinase domain ,hemic and lymphatic diseases ,medicine ,Cancer research ,ROS1 ,Lung cancer ,business ,DISEASE RELAPSE ,Brain metastasis - Abstract
Overcoming resistance to targeted kinase inhibitors is a major clinical challenge in oncology. For 1st and 2nd generation ALK inhibitors acquired resistance due to ALK kinase domain mutations and/or pharmacological drug resistance are major causes for disease relapse. Here, we report the preclinical evaluation of PF-06463922, a potent and brain penetrant ALK/ROS1 inhibitor with sub to low nanomolar cell potency against ALK fusions and all known clinically-acquired resistant mutations. PF-06463922 exhibited marked cytoreductive activity in tumor xenografts driven by various ALK mutants. Furthermore, PF-06463922 led to significant regression of EML4-ALK driven lung cancer brain metastasis and prolonged mouse survival. Compared to other clinically available ALK inhibitors, PF-06463922 is unique in its superior potency against a broad spectrum of acquired ALK mutations, including the highly resistant G1202R mutant and its robust antitumor activity in the brain. Furthermore, PF-06463922 demonstrated remarkable selectivity and safety margins in a variety of preclinical studies. These results suggest that PF-06463922 may be highly effective for the treatment of patients with ALK-driven lung cancers, including those who relapsed on clinically available ALK inhibitors due to ALK secondary mutations and/or brain metastases. Citation Format: Luc Friboulet, Helen Zou, David P. Kodack, Lars D. Engstrom, Qiuhua Li, Melissa West, Ruth W. Tang, Hui Wang, Konstantinos Tsaparikos, Jinwei Wang, Sergei Timofeevski, Dac M. Dinh, Hieu Lam, Justine L. Lam, Shinji Yamazaki, Wenyue Hu, Bhushankumar Patel, Divya Bezwada, Sidra Mahmood, Eugene Lifshits, Timothy Affolter, Patrick B. Lappin, Hovhannes Gukasyan, Nathan Lee, Shibing Deng, Rakesh K. Jain, Ted W. Johnson, Alice T. Shaw, Valeria R. Fantin, Tod Smeal. PF-06463922, a novel next generation ALK/ROS1 inhibitor, overcomes resistance to 1st and 2nd generation ALK inhibitors in pre-clinical models. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 130. doi:10.1158/1538-7445.AM2015-130
- Published
- 2015
- Full Text
- View/download PDF
28. Abstract PR09: Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumor regressions in KRAS-mutant cancer models
- Author
-
Erin M. Coffee, Katherine A. Cheng, Jeffrey A. Engelman, Dora Dias-Santagata, Mari Mino-Kenudson, Jeffrey Settleman, Youngchul Song, Patricia Greninger, Jason T. Godfrey, Cyril H. Benes, Toshi Shioda, Ronald D. Brown, Anurag K. Singh, Kwok-Kin Wong, Eugene Lifshits, Hiromichi Ebi, Travis J. Cohoon, Anthony C. Faber, Ryan B. Corcoran, and Aaron N. Hata
- Subjects
Cancer Research ,Navitoclax ,Oncogene ,biology ,MEK inhibitor ,medicine.medical_treatment ,Cancer ,Bcl-xL ,medicine.disease_cause ,medicine.disease ,Targeted therapy ,chemistry.chemical_compound ,Oncology ,chemistry ,Immunology ,Cancer cell ,Cancer research ,medicine ,biology.protein ,KRAS - Abstract
Although KRAS is the most commonly mutated oncogene in human cancer, KRAS has proven difficult to target pharmacologically, and no effective therapies exist for KRAS-mutant cancers. Recently, there has been evidence that targeted therapy combinations inhibiting multiple downstream effectors of KRAS may be a promising approach for KRAS-mutant cancers. We developed a pooled shRNA-drug screen strategy to identify genes that, when inhibited, cooperate with MEK inhibitors to kill KRAS-mutant cancer cells. The anti-apoptotic BH3 family gene BCL-XL emerged as a top hit through this approach. ABT-263 (navitoclax), a chemical inhibitor that blocks the ability of BCL-XL to bind and inhibit pro-apoptotic proteins, in combination with a MEK inhibitor led to dramatic apoptosis in the vast majority of KRAS-mutant cell lines tested from different tissue types. Mechanistic studies revealed that MEK inhibition led to marked induction of the pro-apoptotic protein BIM in KRAS-mutant cancer cells, but that BIM remained bound and inhibited by BCL-XL. Pharmacologic inhibition of BCL-XL with ABT-263 disrupted this inhibitory complex, allowing BIM to trigger apoptosis. Epithelial differentiation and E-cadherin expression correlated with increased sensitivity to this inhibitor combination across a panel of 30 KRAS-mutant cell lines, while epithelial-to-mesenchymal transition (EMT) correlated with resistance. This combination also caused marked in vivo tumor regressions in three independent KRAS-mutant xenografts and in established lung tumors in two genetically-engineered KRAS-driven lung cancer mouse models. These data support combined BCL-XL/MEK inhibition as a promising therapeutic approach for evaluation in future clinical trials for patients with KRAS-mutant cancers. This abstract is also presented as Poster B19. Citation Format: Ryan B. Corcoran, Katherine A. Cheng, Aaron N. Hata, Anthony C. Faber, Hiromichi Ebi, Erin M. Coffee, Patricia Greninger, Ronald D. Brown, Jason T. Godfrey, Travis J. Cohoon, Youngchul Song, Eugene Lifshits, Toshi Shioda, Dora Dias-Santagata, Anurag Singh, Jeffrey Settleman, Cyril H. Benes, Mari Mino-Kenudson, Kwok-Kin Wong, Jeffrey A. Engelman. Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumor regressions in KRAS-mutant cancer models. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Synthetic Lethal Approaches to Cancer Vulnerabilities; May 17-20, 2013; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(5 Suppl):Abstract nr PR09.
- Published
- 2013
- Full Text
- View/download PDF
29. Abstract 956: Receptor tyrosine kinases, not KRAS, activate PI3K in KRAS mutant colorectal cancers
- Author
-
Lewis C. Cantley, Ryan B. Corcoran, Zhao Chen, David P. Ryan, Eugene Lifshits, Jeffrey Settleman, Cyril H. Benes, Hiromichi Ebi, Kwok-Kin Wong, Youngchul Song, Jeffrey A. Meyerhardt, Anurag K. Singh, and Jeffrey A. Engelman
- Subjects
MAPK/ERK pathway ,Cancer Research ,biology ,Cetuximab ,MEK inhibitor ,Cancer ,medicine.disease_cause ,medicine.disease ,digestive system diseases ,Receptor tyrosine kinase ,Gefitinib ,Oncology ,Cancer research ,medicine ,biology.protein ,KRAS ,neoplasms ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
Introduction: Therapies inhibiting receptor tyrosine kinases (RTKs) are effective when they lead to simultaneous downregulation of phosphoinositide 3-kinase (PI3K)-AKT and mitogen-activated protein kinase (MEK)-ERK signaling. Although cetuximab has demonstrated activity in KRAS wildtype cancers, it remains unknown if RTK inhibition alone can suppress one or both of these pathways in colorectal cancers, especially in cancers with KRAS mutations. Experimental Procedures and Results: Colorectal cancer cells were treated with gefitinib (1 μM) or Cetuximab (10 μg/mL) to compare the consequences of EGFR inhibition on downstream signaling events between KRAS wt (n=6) and mt (n=8) cell lines. In KRAS wt cancers, either cetuximab or gefitinib led to loss of ERK phosphorylation, but had only a modest effect on PI3K signaling. However, in most KRAS mutant cancers, EGFR inhibition minimally impacted ERK signaling and had neglible effects on PI3K signaling. To determine if KRAS constitutively activates ERK and PI3K signaling in KRAS mutant colorectal cancers, we utilized shRNA in eight KRAS mutant cancer cell lines and abruptly turned off KRAS expression in an vivo transgenic model. In all models, the phosphorylation of ERK was markedly decreased by KRAS knockdown, but this was not associated with downregulation of PI3K signaling. To determine how PI3K was activated in KRAS mutant cancers, we immunoprecipitated the p85 regulatory subunit and identified that IRS proteins co-precipitated with PI3K in KRAS mutant cancer. These complexes were disrupted by an IGF-IR inhibitor, NVP-AEW541, but not by gefitinib, and accordingly, NVP-AEW541 led to loss of AKT phosphorylation. Of the eight KRAS mutant cell lines, we found p85/IRS-1 or p85/IRS-2 complexes in 7 cell lines. We also observed that a monoclonal anti-IGF-IR antibody R1507 (18 mg/kg twice a week) downregulated PI3K signaling in SW837 xenografts in vivo. Biochemical assessment of patient specimens with KRAS mutations also suggested that PI3K was regulated primarily by IGF-IR. Importantly, by combining RTK inhibitors with MEK inhibitors in KRAS mutant cancers, both the PI3K and ERK pathways were concomitantly downregulated leading to marked growth suppression and robust apoptosis. Furthermore, combining R1507 (18 mg/kg twice a week) with a MEK inhibitor (AZD6244 25 mg/kg twice daily) induced marked tumor regressions of a KRAS mutant colorectal cancer in vivo. Conclusions: In this study, we provide evidence that KRAS does not drive PI3K signaling in KRAS mutant cancers, and demonstrate that specific RTK inhibition potently suppresses PI3K signaling in vitro and in vivo. These findings provide a framework for utilizing RTK inhibitors in the treatment of KRAS mutant colorectal cancers. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 956. doi:10.1158/1538-7445.AM2011-956
- Published
- 2011
- Full Text
- View/download PDF
30. Preexistence and Clonal Selection of MET Amplification in EGFR Mutant NSCLC
- Author
-
Sara Akhavanfard, Alexa B. Turke, Tony Mok, Youngchul Song, James G. Christensen, Eugene Lifshits, Pasi A. Jänne, Luca Toschi, Neal I. Lindeman, Yi-Long Wu, Beow Y. Yeap, Dora Dias-Santagata, Carly Murphy, Marzia Capelletti, Kreshnik Zejnullahu, Andrew H. Rogers, Lecia V. Sequist, Yun Xiao, A. John Iafrate, Jeffrey A. Engelman, and Charles Lee
- Subjects
Cancer Research ,Lung Neoplasms ,Gene Expression ,Antineoplastic Agents ,CELLCYCLE ,Biology ,Polymerase Chain Reaction ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,ERBB3 ,Receptors, Growth Factor ,Autocrine signalling ,Lung cancer ,Protein kinase B ,PI3K/AKT/mTOR pathway ,In Situ Hybridization, Fluorescence ,030304 developmental biology ,Quinazolinones ,0303 health sciences ,Hepatocyte Growth Factor ,Gene Amplification ,Cell Biology ,Cell cycle ,Proto-Oncogene Proteins c-met ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,ErbB Receptors ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Hepatocyte growth factor ,medicine.drug ,Signal Transduction - Abstract
SummaryMET amplification activates ERBB3/PI3K/AKT signaling in EGFR mutant lung cancers and causes resistance to EGFR kinase inhibitors. We demonstrate that MET activation by its ligand, HGF, also induces drug resistance, but through GAB1 signaling. Using high-throughput FISH analyses in both cell lines and in patients with lung cancer, we identify subpopulations of cells with MET amplification prior to drug exposure. Surprisingly, HGF accelerates the development of MET amplification both in vitro and in vivo. EGFR kinase inhibitor resistance, due to either MET amplification or autocrine HGF production, was cured in vivo by combined EGFR and MET inhibition. These findings highlight the potential to prospectively identify treatment naive, patients with EGFR-mutant lung cancer who will benefit from initial combination therapy.
- Full Text
- View/download PDF
31. Synthetic Lethal Interaction of Combined BCL-XL and MEK Inhibition Promotes Tumor Regressions in KRAS Mutant Cancer Models
- Author
-
Eugene Lifshits, Youngchul Song, Mari Mino-Kenudson, Anurag K. Singh, Patricia Greninger, Jeffrey Settleman, Ryan B. Corcoran, Travis J. Cohoon, Ronald D. Brown, Toshi Shioda, Erin M. Coffee, Anthony C. Faber, Katherine A. Cheng, Jeffrey A. Engelman, Kenneth E. Hung, Kwok-Kin Wong, Hiromichi Ebi, Dora Dias-Santagata, Aaron N. Hata, Cyril H. Benes, and Jason T. Godfrey
- Subjects
Cancer Research ,Mutant ,bcl-X Protein ,Antineoplastic Agents ,Bcl-xL ,Biology ,medicine.disease_cause ,Proto-Oncogene Proteins p21(ras) ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,medicine ,Animals ,Humans ,neoplasms ,030304 developmental biology ,Sulfonamides ,0303 health sciences ,Aniline Compounds ,Navitoclax ,Oncogene ,MEK inhibitor ,Cancer ,Cell Biology ,MAP Kinase Kinase Kinases ,medicine.disease ,Molecular biology ,digestive system diseases ,respiratory tract diseases ,3. Good health ,chemistry ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,Benzimidazoles ,KRAS ,Drug Screening Assays, Antitumor - Abstract
SummaryKRAS is the most commonly mutated oncogene, yet no effective targeted therapies exist for KRAS mutant cancers. We developed a pooled shRNA-drug screen strategy to identify genes that, when inhibited, cooperate with MEK inhibitors to effectively treat KRAS mutant cancer cells. The anti-apoptotic BH3 family gene BCL-XL emerged as a top hit through this approach. ABT-263 (navitoclax), a chemical inhibitor that blocks the ability of BCL-XL to bind and inhibit pro-apoptotic proteins, in combination with a MEK inhibitor led to dramatic apoptosis in many KRAS mutant cell lines from different tissue types. This combination caused marked in vivo tumor regressions in KRAS mutant xenografts and in a genetically engineered KRAS-driven lung cancer mouse model, supporting combined BCL-XL/MEK inhibition as a potential therapeutic approach for KRAS mutant cancers.
- Full Text
- View/download PDF
32. Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab
- Author
-
Osamu Maenishi, Yasuhito Fujisaka, John Souglakos, Kazuto Nishio, Koji Takeda, Dalia Ercan, Kimio Yonesaka, Hiroyuki Itoh, Pasi A. Jänne, Jason Sun, Yonggon Cho, Minoru Takada, Ramesh A. Shivdasani, Kreshnik Zejnullahu, Toshio Shimizu, Juliet Philips, Marileila Varella-Garcia, Kazuhiko Nakagawa, Masayuki Takeda, Annarita Destro, Masahiro Fukuoka, Kiyotaka Okuno, Eugene Lifshits, Federico Cappuzzo, Jeffrey A. Engelman, Isamu Okamoto, Jeffrey Swanson, Taroh Satoh, Koichi Taira, Massimo Roncalli, Takafumi Okabe, and Andrew H. Rogers
- Subjects
Receptor, ErbB-2 ,Colorectal cancer ,Neuregulin-1 ,Transplantation, Heterologous ,Cetuximab ,Antineoplastic Agents ,Antibodies, Monoclonal, Humanized ,medicine.disease_cause ,Article ,Mice ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,ERBB3 ,Epidermal growth factor receptor ,Neuregulin 1 ,skin and connective tissue diseases ,neoplasms ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,biology ,Antibodies, Monoclonal ,General Medicine ,medicine.disease ,digestive system diseases ,ErbB Receptors ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,Neuregulin ,KRAS ,Signal transduction ,Neoplasm Transplantation ,Signal Transduction ,medicine.drug - Abstract
Cetuximab, an antibody directed against the epidermal growth factor receptor, is an effective clinical therapy for patients with colorectal, head and neck, and non-small cell lung cancer, particularly for those with KRAS and BRAF wild-type cancers. Treatment in all patients is limited eventually by the development of acquired resistance, but little is known about the underlying mechanism. Here, we show that activation of ERBB2 signaling in cell lines, either through ERBB2 amplification or through heregulin up-regulation, leads to persistent extracellular signal-regulated kinase 1/2 signaling and consequently to cetuximab resistance. Inhibition of ERBB2 or disruption of ERBB2/ERBB3 heterodimerization restores cetuximab sensitivity in vitro and in vivo. A subset of colorectal cancer patients who exhibit either de novo or acquired resistance to cetuximab-based therapy has ERBB2 amplification or high levels of circulating heregulin. Collectively, these findings identify two distinct resistance mechanisms, both of which promote aberrant ERBB2 signaling, that mediate cetuximab resistance. Moreover, these results suggest that ERBB2 inhibitors, in combination with cetuximab, represent a rational therapeutic strategy that should be assessed in patients with cetuximab-resistant cancers.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.